quantitative phase imaging
Recently Published Documents


TOTAL DOCUMENTS

612
(FIVE YEARS 257)

H-INDEX

37
(FIVE YEARS 8)

2022 ◽  
Vol 150 ◽  
pp. 106833
Author(s):  
Shengyu Lu ◽  
Yong Tian ◽  
Qinnan Zhang ◽  
Xiaoxu Lu ◽  
Jindong Tian

2021 ◽  
Vol 13 (4) ◽  
pp. 91
Author(s):  
Arkadiusz Kuś ◽  
Wojciech Krauze ◽  
Małgorzata Kujawińska

In this paper we briefly present the history and outlook on the development of two seemingly distant techniques which may be brought close together with a unified theoretical model described as common k-space theory. This theory also known as the Fourier diffraction theorem is much less common in optical coherence tomography than its traditional mathematical model, but it has been extensively studied in digital holography and, more importantly, optical diffraction tomography. As demonstrated with several examples, this link is one of the important factors for future development of both techniques. Full Text: PDF ReferencesN. Leith, J. Upatnieks, "Reconstructed Wavefronts and Communication Theory", J. Opt. Soc. Am. 52(10), 1123 (1962). CrossRef Y. Park, C. Depeursinge, G. Popescu, "Quantitative phase imaging in biomedicine", Nat. Photonics 12, 578 (2018). CrossRef D. Huang et al., "Optical Coherence Tomography", Science 254(5035), 1178 (1991). CrossRef D. P. Popescu, C. Flueraru, S. Chang, J. Disano, S. Sherif, M.G. Sowa, "Optical coherence tomography: fundamental principles, instrumental designs and biomedical applications", Biophys. Rev. 3(3), 155 (2011). CrossRef M. Wojtkowski, V. Srinivasan, J.G. Fujimoto, T. Ko, J.S. Schuman, A. Kowalczyk, J.S. Duker, "Three-dimensional Retinal Imaging with High-Speed Ultrahigh-Resolution Optical Coherence Tomography", Ophthalmology 112(10), 1734 (2005). CrossRef K.C. Zhou, R. Qian, A.-H. Dhalla, S. Farsiu, J.A. Izatt, "Unified k-space theory of optical coherence tomography", Adv. Opt. Photon. 13(2), 462 (2021). CrossRef A.F. Fercher, C.K. Hitzenberger, G. Kamp, S.Y. El-Zaiat, "Measurement of intraocular distances by backscattering spectral interferometry", Opt. Comm. 117(1-2), 43 (1995). CrossRef E. Wolf, "Determination of the Amplitude and the Phase of Scattered Fields by Holography", J. Opt. Soc. Am. 60(1), 18 (1970). CrossRef E. Wolf, "Three-dimensional structure determination of semi-transparent objects from holographic data", Opt. Comm. 1(4), 153 (1969). CrossRef V. Balasubramani et al., "Roadmap on Digital Holography-Based Quantitative Phase Imaging", J. Imaging 7(12), 252 (2021). CrossRef A. Kuś, W. Krauze, P.L. Makowski, M. Kujawińska, "Holographic tomography: hardware and software solutions for 3D quantitative biomedical imaging (Invited paper)", ETRI J. 41(1), 61 (2019). CrossRef A. Kuś, M. Dudek, M. Kujawińska, B. Kemper, A. Vollmer, "Tomographic phase microscopy of living three-dimensional cell cultures", J. Biomed. Opt. 19(4), 46009 (2014). CrossRef O. Haeberlé, K. Belkebir, H. Giovaninni, A. Sentenac, "Tomographic diffractive microscopy: basics, techniques and perspectives", J. Mod. Opt. 57(9), 686 (2010). CrossRef B. Simon et al., "Tomographic diffractive microscopy with isotropic resolution", Optica 4(4), 460 (2017). CrossRef B.A. Roberts, A.C. Kak, "Reflection Mode Diffraction Tomography", Ultrason. Imag. 7, 300 (1985). CrossRef M. Sarmis et al., "High resolution reflection tomographic diffractive microscopy", J. Mod. Opt. 57(9), 740 (2010). CrossRef L. Foucault et al., "Versatile transmission/reflection tomographic diffractive microscopy approach", J. Opt. Soc. Am. A 36(11), C18 (2019). CrossRef W. Krauze, P. Ossowski, M. Nowakowski, M. Szkulmowski, M. Kujawińska, "Enhanced QPI functionality by combining OCT and ODT methods", Proc. SPIE 11653, 116530B (2021). CrossRef E. Mudry, P.C. Chaumet, K. Belkebir, G. Maire, A. Sentenac, "Mirror-assisted tomographic diffractive microscopy with isotropic resolution", Opt. Lett. 35(11), 1857 (2010). CrossRef P. Hosseini, Y. Sung, Y. Choi, N. Lue, Z. Yaqoob, P. So, "Scanning color optical tomography (SCOT)", Opt. Expr. 23(15), 19752 (2015). CrossRef J. Jung, K. Kim, J. Yoon, Y. Park, "Hyperspectral optical diffraction tomography", Opt. Expr. 24(3), 1881 (2016). CrossRef T. Zhang et al., Biomed. "Multi-wavelength multi-angle reflection tomography", Opt. Expr. 26(20), 26093 (2018). CrossRef R.A. Leitgeb, "En face optical coherence tomography: a technology review [Invited]", Biomed. Opt. Expr. 10(5), 2177 (2019). CrossRef J.F. de Boer, R. Leitgeb, M. Wojtkowski, "Twenty-five years of optical coherence tomography: the paradigm shift in sensitivity and speed provided by Fourier domain OCT [Invited]", Biomed. Opt. Expr. 8(7), 3248 (2017). CrossRef T. Anna, V. Srivastava, C. Shakher, "Transmission Mode Full-Field Swept-Source Optical Coherence Tomography for Simultaneous Amplitude and Quantitative Phase Imaging of Transparent Objects", IEEE Photon. Technol. Lett. 23(11), 899 (2011). CrossRef M.T. Rinehart, V. Jaedicke, A. Wax, "Quantitative phase microscopy with off-axis optical coherence tomography", Opt. Lett. 39(7), 1996 (2014). CrossRef C. Photiou, C. Pitris, "Dual-angle optical coherence tomography for index of refraction estimation using rigid registration and cross-correlation", J. Biomed. Opt. 24(10), 1 (2019). CrossRef Y. Zhou, K.K.H. Chan, T. Lai, S. Tang, "Characterizing refractive index and thickness of biological tissues using combined multiphoton microscopy and optical coherence tomography", Biomed. Opt. Expr. 4(1), 38 (2013). CrossRef K.C. Zhou, R. Qian, S. Degan, S. Farsiu, J.A. Izatt, "Optical coherence refraction tomography", Nat. Photon. 13, 794 (2019). CrossRef


2021 ◽  
Author(s):  
Daniele Pirone ◽  
Joowon Lim ◽  
Francesco Merola ◽  
Lisa Miccio ◽  
Martina Mugnano ◽  
...  

Quantitative Phase Imaging (QPI) has gained popularity because it can avoid the staining step, which in some cases is difficult or impossible. However, QPI does not provide the well-known specificity to various parts of the cell (e.g., organelles, membrane). Here we show a novel computational segmentation method based on statistical inference that bridges the gap between the specificity of Fluorescence Microscopy (FM) and the label-free property of QPI techniques to identify the cell nucleus. We demonstrate application to stain-free cells reconstructed through the holographic learning and in flow cyto-tomography modality. In particular, by means of numerical simulations and two cancer cell lines, we demonstrate that the nucleus-like regions can be accurately distinguished within the stain-free tomograms. We show that our experimental results are consistent with confocal FM data and microfluidic cytofluorimeter outputs. This is a significant step towards extracting the three-dimensional (3D) intracellular specificity directly from the phase-contrast data in a typical flow cytometry configuration.


Sensors ◽  
2021 ◽  
Vol 21 (23) ◽  
pp. 8021
Author(s):  
Raul Castaneda ◽  
Carlos Trujillo ◽  
Ana Doblas

The conventional reconstruction method of off-axis digital holographic microscopy (DHM) relies on computational processing that involves spatial filtering of the sample spectrum and tilt compensation between the interfering waves to accurately reconstruct the phase of a biological sample. Additional computational procedures such as numerical focusing may be needed to reconstruct free-of-distortion quantitative phase images based on the optical configuration of the DHM system. Regardless of the implementation, any DHM computational processing leads to long processing times, hampering the use of DHM for video-rate renderings of dynamic biological processes. In this study, we report on a conditional generative adversarial network (cGAN) for robust and fast quantitative phase imaging in DHM. The reconstructed phase images provided by the GAN model present stable background levels, enhancing the visualization of the specimens for different experimental conditions in which the conventional approach often fails. The proposed learning-based method was trained and validated using human red blood cells recorded on an off-axis Mach–Zehnder DHM system. After proper training, the proposed GAN yields a computationally efficient method, reconstructing DHM images seven times faster than conventional computational approaches.


2021 ◽  
Author(s):  
Edward R Polanco ◽  
Tarek E Moustafa ◽  
Andrew Butterfield ◽  
Sandra D Scherer ◽  
Emilio Cortes-Sanchez ◽  
...  

Quantitative phase imaging (QPI) measures the growth rate of individual cells by quantifying changes in mass versus time. Here, we use the breast cancer cell lines MCF-7, BT-474, and MDA-MB-231 to validate QPI as a multiparametric approach for determining response to single-agent therapies. Our method allows for rapid determination of drug sensitivity, cytotoxicity, heterogeneity, and time of response for up to 100,000 individual cells or small clusters in a single experiment. We find that QPI EC50 values are concordant with CellTiter-Glo (CTG), a gold standard metabolic endpoint assay. In addition, we apply multiparametric QPI to characterize cytostatic/cytotoxic and rapid/slow responses and track the emergence of resistant subpopulations. Thus, QPI reveals dynamic changes in response heterogeneity in addition to average population responses, a key advantage over endpoint viability or metabolic assays. Overall, multiparametric QPI reveals a rich picture of cell growth by capturing the dynamics of single-cell responses to candidate therapies.


Sign in / Sign up

Export Citation Format

Share Document